EP1843779A2 - Zusammensetzungen mit antiangiogenem phscn-peptid - Google Patents

Zusammensetzungen mit antiangiogenem phscn-peptid

Info

Publication number
EP1843779A2
EP1843779A2 EP06720021A EP06720021A EP1843779A2 EP 1843779 A2 EP1843779 A2 EP 1843779A2 EP 06720021 A EP06720021 A EP 06720021A EP 06720021 A EP06720021 A EP 06720021A EP 1843779 A2 EP1843779 A2 EP 1843779A2
Authority
EP
European Patent Office
Prior art keywords
composition
peptide
acid
analogue
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP06720021A
Other languages
English (en)
French (fr)
Inventor
Andrew P Mazar
Hashem Heiati
Jay Schrier
Ming Li
Scott Harris
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tactic Pharma LLC
Original Assignee
Attenuon LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Attenuon LLC filed Critical Attenuon LLC
Publication of EP1843779A2 publication Critical patent/EP1843779A2/de
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/16Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
    • A61K47/18Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
    • A61K47/183Amino acids, e.g. glycine, EDTA or aspartame
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention is directed to compositions comprising improved formulations of the Cys-containing anti-angiogenic peptide Pro-His-Ser-Cys-Asn that prevent degradation and spontaneous oxidative dimerization or oligomerization of the peptide.
  • angiogenesis i.e., aberrant vascularization
  • normal tissue does not require angiogenesis except under specialized circumstances such as wound repair, the proliferation of the uterine internal lining during the menstrual cycle, etc. Accordingly, a requirement for angiogenesis is a significant difference between tumor/cancerous tissue and normal tissue.
  • Tumor angiogenesis can be initiated by cytokines such as vascular endothelial growth factor (VEGF) and/or a fibroblast growth factor (FGF), which bind to specific receptors on endothelial cells ("ECs") in the local vasculature, under hypoxic conditions.
  • VEGF vascular endothelial growth factor
  • FGF fibroblast growth factor
  • the activated ECs secrete enzymes which remodel the associated tissue matrix and modulate expression of adhesion molecules such as integrins. Following matrix degradation, ECs proliferate and migrate toward the tumor, which results in the generation and maturation of new blood vessels.
  • protein fragments such as endostatin, kringle 5 and PEX, which inhibit angiogenesis, are produced by degradation of matrix proteins (O'Reilly et ah, Cell 1997, 8#:277-85; O'Reilly et ah, Cell, 1994, 79:315-28; Brooks etah, Cell, 1998, 92:391-400). Accordingly, these protein fragments may inhibit neoangiogenesis, thus preventing tumor growth and metastasis.
  • protein fragments have significant drawbacks associated with their use. They are difficult and expensive to produce in large quantities, show poor pharmacological properties, are susceptible to degradation, etc.
  • One approach has been to identify short peptide fragments of these larger proteins or of the longer fragments or subunits, which shorter peptides retain a significant portion of the anti-angiogenic activity of the parent protein.
  • novel peptides are needed to fully explore the potential of peptides in preventing angiogenesis and detecting aberrant vascularization.
  • the novel peptides may have longer plasma half-lives, be more resistant to degradation, have increased bio-availability, higher affinity, greater selectivity, etc. in comparison to peptides described in the art (Livant, U.S. Pats. No. 6,001,965 and 6,472,369).
  • Such novel peptides may be effective in inhibiting cell migration, invasion and proliferation and treating or preventing various diseases associated with undesired angiogenesis and aberrant vascularization.
  • One approach to improving the activity profile of an anti-angiogenic peptide is to exploit the method of its formulation as a means to extend shelf life and enhance the resistance to degradation.
  • peptides that include Cys it is also important to prevent spontaneous oxidative dimerization or higher degrees of oligomerization or polymerization through disulfide bond formation.
  • the present invention is directed to such improved formulations for anti-angiogenic peptide Ac-PHSCN-NH 2 and its anti-angiogenic derivatives.
  • compositions comprising improved formulations for Cys-containing peptides and salts thereof that preserve the biological/biochemical potency of the peptides, particularly by inhibiting disulfide bond formation that leads to undesired dimerization.
  • Acid addition salts OfAc-PHSCN-NH 2 useful in the formulations of the present invention are described in co-pending applications by Trenansky et ah, both entitled “Acid Addition Salts of Ac-PHSCN-NH 2 ", United States Provisional Application Serial No. 60/649,308, filed February 1, 2005, and International Application No. (to be assigned), filed February 1, 2006 (Attorney Docket No. 9715-022-228), each of which is incorporated by reference herein in its entirety.
  • the above application discloses acid addition salts OfAc-PHSCN-NH 2 , methods of making acid addition salts OfAc-PHSCN-NH 2 , pharmaceutical compositions of acid addition salts OfAc-PHSCN-NH 2 , methods of using acid addition salts OfAc-PHSCN-NH 2 and pharmaceutical compositions thereof to treat diseases associated with angiogenesis and aberrant vascularization and methods of preventing degradation of Ac-PHSCN-NH 2 by salt formation.
  • the novel formulations of the present application include those using the new salts described by Ternansky et at, supra.
  • the present invention is thus directed to a formulation comprising a peptide Pro-His-Ser- Cys-Asn (PHSCN), an analogue thereof of a salt of the peptide or analogue, and at least one additional compound that stabilizes the peptide or analogue against spontaneous tandem dimerization or higher oligomerization.
  • PHSCN Pro-His-Ser- Cys-Asn
  • the peptide is capped at both termini, with an acetyl group at the N-terminus and amide group at the C-terminus.
  • the additional compound inhibits, prevents or reverses disulfide bond formation between sulfhydryl groups of Cys residues.
  • the additional compound is a biocompatible acid buffer with a pK of about 5.
  • the pH of the solution is greater than about 3.0 and less than about, or equal to, 7.5.
  • a preferred acid buffer is citrate, acetate or 2-(N-morpho- lino)ethanesulfonic acid (MES).
  • the acid buffer is preferably citrate at a concentration of about 25 mM.
  • the buffer may be supplemented with glycine as an excipient and bulking agent, preferably at a concentration of about 50 mg/ml.
  • the buffer may comprise citrate and acetate and may also comprise Tris.
  • the above formulation comprises (i) the peptide, capped peptide or analogue, (ii) about 50 mM citrate, and (iii) about 50 mg/ml glycine.
  • a preferred embodiment of the formulation is in a container or vial in lyophilized form having 100 mg peptide, capped peptide, salt or analogue, 50 mM citrate, 50 mg/ml glycine lyophilized from 2 mL of a pH 5.0 solution.
  • the above formulation may further comprise or more reducing agents; preferred agents are dithiothreitol ⁇ -mercaptoethanol, or glutathione.
  • the concentration of the reducing agent or agents does not exceed about 10 mM.
  • the above formulation may comprise a non-thiol biocompatible anti-oxidant.
  • the formulation may comprise a lyoprotectant present in an lyoprotecting amount, for example, about 50-600 mole lyoprotectant: 1 mole peptide.
  • the lyoprotectant is one or more sugars, one or more amino acids, one or more methylamine, one or more lyotropic salts, and/or one or more polyols.
  • a lyoprotectant present in an lyoprotecting amount, for example, about 50-600 mole lyoprotectant: 1 mole peptide.
  • the lyoprotectant is one or more sugars, one or more amino acids, one or more methylamine, one or more lyotropic salts, and/or one or more polyols.
  • the sugar is sucrose or trehalose;
  • the amino acid is monosodium glutamate or histidine;
  • the methylamine is betaine;
  • the lyotropic salt is magnesium sulfate; and
  • the polyol is a trihydric or higher sugar alcohol.
  • the formulation above may comprise one or more polyols selected from the group consisting of glycerin, erythritol, glycerol, arabitol, xylitol, sorbitol, mannitol, propylene glycol, and a combination thereof.
  • the lyoprotectant is a non-reducing sugar, preferably trehalose or sucrose.
  • the peptide is preferably capped at its N- and C-termini, most preferably the N-terminal cap is an acyl group and the C terminal cap is an amide group such that the peptide terminates in CO-NH 2 .
  • the formulation above is preferably sterile and formulated for in vivo administration.
  • the present invention includes an article of manufacture or kit comprising (a) a container which contains a formulation as above, in solution or in lyophilized form;
  • the article or kit may further comprise one or more of (i) another buffer, (ii) a diluent, (iii) a filter, (iv) a needle, or (v) a syringe.
  • the container is preferably a bottle, a vial, a syringe or test tube; it may be a multi-use container, the formulation is preferably lyophilized.
  • the present invention is directed to a method of inhibiting angiogenesis in a subject, comprising administering to the subject a formulation as above , wherein the peptide, analogue or salt is administered in an anti-angiogenic effective amount.
  • Such inhibition of angiogenesis is exploited in a method for the treatment of cancer and or Crohn's disease.
  • the invention provides use of a composition as above in a medicament for administration to a subject to inhibit undesired angiogenesis, such as in the treatment of cancer or Crohn's disease. Also included is use of a composition as above in the manufacture of a medicament for administration to a subject to inhibit undesired angiogenesis and, thereby, to treat cancer and Crohn's disease.
  • PHSCN most preferably, the terminally capped pentapeptide, Acetyl-PHSCN-NH 2 , and various salts thereof, and or analogues of capped or uncapped PHSCN and salts of the derivatives and analogues.
  • An "analogue" of PHSCN refers to a molecule, natural or non- natural, that is structurally and/or functionally substantially similar to PHSCN. Examples include conservative amino acid substitution variants, addition variants of not more than about 20 residues (to the exclusion of native polypeptides or their structural domains), and chemically modified peptides. Other analogues are peptidomimetics and aptamers.
  • the peptide to be formulated is preferably pure, or essentially pure and, desirably, essentially homogeneous ⁇ i.e., free from contaminating peptides or proteins, etc.)
  • "Essentially pure” means a peptide preparation wherein at least 90% by weight is the peptide based on total weight of the preparation, preferably at least 95% by weight.
  • An "essentially homogeneous” preparation means a peptide preparation comprising at least 99% by weight of peptide, based on total weight of the peptide in the preparation.
  • the PHSCN peptide is preferably capped at its N and C termini with an acyl (abbreviated “Ac”) -and an amido (abbreviated “Am”) group, respectively, for example acetyl (CH 3 CO-) at the N terminus and amido (CO-NH 2 ) at the C terminus.
  • Ac acyl
  • Am amido
  • N-terminal capping functions preferably in a linkage to the terminal amino group, is contemplated, for example: formyl;
  • C 1-10 alkanoyl such as acetyl, propionyl, butyryl
  • C 1-1O alkenoyl such as hex-3-enoyl
  • C 1-10 alkynoyl having from 1 to 10 carbon atoms, such as hex-5-ynoyl; aroyl, such as benzoyl or 1-naphthoyl; heteroaroyl, such as 3-pyrroyl or 4-quinoloyl; alkylsulfonyl, such as methanesulfonyl; arylsulfonyl, such as benzenesulfonyl or sulfanilyl; heteroarylsulfonyl, such as pyridine-4-sulfonyl;
  • substituted alkynoyl such as 3-hydroxy-hex-5-ynoyl; substituted aroyl, such as 4-chlorobenzoyl or 8-hydroxy-naphth-2-oyl; substituted heteroaroyl, such as 2,4-dioxo-l,2,3,4-tetrahydro-3-methyl-quinazolin-6-oyl; substituted alkylsulfonyl, such as 2-aminoethanesulfonyl; substituted arylsulfonyl, such as 5-dimethylamino-l-naphthalenesulfonyl; substituted heteroarylsulfonyl, such as l-methoxy-6-isoquinolinesulfonyl; carbamoyl or thiocarbamoyl; substituted carbamoyl (R'-NH-CO) or substituted thiocarbamoyl (R' -NH-CS) wherein R
  • the C-terminal capping function can either be in an amide or ester bond with the terminal
  • NR R wherein R and R 2 may be independently drawn from the following group: hydrogen;
  • C 1-I0 alkyl such as methyl, ethyl, isopropyl
  • Ci-io alkenyl such as prop-2-enyl
  • Ci-io alkynyl such as prop-2-ynyl
  • Ci-io substituted alkyl such as hydroxyalkyl, alkoxyalkyl, mercaptoalkyl, alkylthioalkyl, halogenoalkyl, cyanoalkyl, aminoalkyl, alkylaminoalkyl, dialkylaminoalkyl, alkanoylalkyl, carboxyalkyl, carbamoylalkyl;
  • C 1-IO substituted alkenyl such as hydroxyalkenyl, alkoxyalkenyl, mercaptoalkenyl, alkylthioalkenyl, halogenoalkenyl, cyanoalkenyl, aminoalkenyl, alkylaminoalkenyl, dialkylaminoalkenyl, alkanoylalkenyl, carboxyalkenyl, carbamoylalkenyl;
  • Ci-io substituted alkynyl such as hydroxyalkynyl, alkoxyalkynyl, mercaptoalkynyl, alkylthioalkynyl, halogenoalkynyl, cyanoalkynyl, aminoalkynyl, alkylaminoalkynyl, dialkylaminoalkynyl, alkanoylalkynyl, carboxyalkynyl, carbamoylalkynyl;
  • C 1 - I o aroylalkyl such as phenacyl or 2-benzoylethyl; aryl, such as phenyl or 1-naphthyl; heteroaryl, such as 4-quinolyl;
  • C 1- IO alkanoyl such as acetyl or butyryl; aroyl, such as benzoyl; heteroaroyl, such as 3-quinoloyl;
  • R' and R" are independently hydrogen, alkyl, aryl, heteroaryl, acyl, aroyl, sulfonyl, sulfinyl, or SO 2 -R' ' ' or SO-R' ' ' where R" ' is substituted or unsubstituted alkyl, aryl, heteroaryl, alkenyl, or alkynyl.
  • Capping functions that provide for an ester bond are designated as OR, wherein R may be: alkoxy; aryloxy; heteroaryloxy; aralkyloxy; heteroaralkyloxy; substituted alkoxy; substituted aryloxy; substituted heteroaryloxy; substituted aralkyloxy; or substituted heteroaralkyloxy.
  • Either the N- terminal or the C-terminal capping function, or both, may be of such structure that the capped molecule functions as a prodrug (a pharmacologically inactive derivative of the parent drug molecule) that undergoes spontaneous or enzymatic transformation within the body in order to release the active drug and that has improved delivery properties over the parent drug molecule (Bundgaard H, Ed: Design of Prodrugs, Elsevier, Amsterdam, 1985).
  • a “lyoprotectant” is a molecule which, when combined with a protein or peptide of interest, significantly prevents or reduces chemical and/or physical instability of the protein or peptide upon lyophilization and subsequent storage.
  • exemplary lyoprotectants include sugars such as sucrose or trehalose; an amino acid such as monosodium glutamate or histidine; a methylamine such as betaine; a lyotropic salt such as magnesium sulfate; a polyol such as trihydric or higher sugar alcohols, e.g., glycerin, erythritol, glycerol, arabitol, xylitol, sorbitol, and mannitol; propylene glycol; polyethylene glycol; pluronics; and combinations thereof.
  • the preferred lyoprotectant is a non-reducing sugar, such as trehalose or sucrose.
  • the lyoprotectant is added to the pre-lyophilized formulation in a "lyoprotecting amount" which means that, following lyophilization of the peptide in the presence of the lyoprotecting amount of the lyoprotectant, the peptide essentially retains its physical and chemical stability and integrity upon lyophilization and storage.
  • the "diluent" of interest is one which is pharmaceutically acceptable (safe and non-toxic for administration to a human) and is useful for the preparation of a reconstituted formulation.
  • exemplary diluents include sterile water, bacteriostatic water for injection (BWFI), a pH buffered solution (e.g. phosphate-buffered saline), sterile saline solution, Ringer's solution or dextrose solution.
  • a "preservative" is a compound which can be added to the diluent to essentially reduce bacterial action in the reconstituted formulation, thus facilitating the production of a multi-use reconstituted formulation, for example.
  • potential preservatives include octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkoniurn chloride (a mixture of alkylbenzyldimethylammonium chlorides in which the alkyl groups are long-chain compounds), and benzethonium chloride.
  • preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol.
  • aromatic alcohols such as phenol, butyl and benzyl alcohol
  • alkyl parabens such as methyl or propyl paraben
  • catechol resorcinol
  • cyclohexanol 3-pentanol
  • m-cresol m-cresol
  • a “bulking agent” is a compound which adds mass to the lyophilized mixture and contributes to the physical structure of the lyophilized cake (e.g., facilitates the production of an essentially uniform lyophilized cake which maintains an open pore structure).
  • Exemplary bulking agents include mannitol, glycine, polyethylene glycol and xorbitol.
  • One important purpose of the present formulation is to stabilize these peptide or analogue against spontaneous dimerization.
  • This is preferably accomplished by formulating the peptides at low pH that will prevent disulfide bond formation between two Cys residues.
  • Such acid buffers are preferably biocompatible. Examples include citrate, acetate, 2-(N-morpholino)ethanesulfonic acid (MES), or any similar buffer with a pK of about 5, wherein in the presence of the buffer, the pH of the solution is ⁇ 7.5 but preferably not below 3.0.
  • a preferred acidic formulation comprises citrate, preferably at about 25mM.
  • the buffer is preferably supplemented with glycine (GIy) as an excipient and bulking agent.
  • GIy glycine
  • a preferred concentration is about 50 mg/ml GIy. Another advantage of GIy is that it is an accepted excipient for intravenous infusion or injection in humans.
  • the present invention includes, in addition to lyophilized compositions, stabilized liquid pharmaceutical compositions comprising a peptide as disclosed herein, preferably capped, whose effectiveness as a therapeutically active component is normally compromised during storage in liquid formulations as a result of dimerization and higher order oligomerization, aggregation, etc.
  • the stabilized liquid pharmaceutical composition comprises an amount of an amino acid base sufficient to decrease aggregate formation during storage, where the amino acid base is an amino acid or a combination of amino acids, where any given amino acid is present either in its free base form or in its salt form. It is understood that the composition comprises a buffering agent to maintain pH of the liquid composition within an acceptable range for stability of the peptide, where the buffering agent is an acid substantially free of its salt form, an acid in its salt form, or a mixture of an acid and its salt form.
  • the amino acid base serves to stabilize the peptide against aggregate formation during storage of the liquid pharmaceutical composition, while use of an acid buffering agent (in either of it forms) results in a liquid composition having an osmolarity that is nearly isotonic.
  • the liquid pharmaceutical composition may additionally incorporate other stabilizing agents, more particularly methionine, a nonionic surfactant such as polysorbate 80, and EDTA, to further increase stability of the peptide.
  • Such liquid pharmaceutical compositions are said to be stabilized, as addition of amino acid base in combination with an acid substantially free of its salt form, an acid in its salt form, or a mixture of an acid and its salt form, results in the increased storage stability relative to liquid compositions formulated in the absence of the combination of these two components.
  • Methods for increasing the stability of the peptide in a liquid pharmaceutical composition comprise incorporating into the liquid composition an amount of an amino acid base sufficient to decrease aggregate formation of the polypeptide during storage, and a buffering agent that is an acid substantially free of its salt form, an acid in its salt form, or a mixture of an acid and its salt form.
  • the acid addition salts Of Ac-PHSCN-NH 2 may be formed from both organic and inorganic acids.
  • Exemplary organic acids include generally, carboxylic acids and sulfonic acids, such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, glycolic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, fumaric acid, oxalic acid, lactic acid, 4- chlorobenzenesulfonic acid, 2-na
  • the acid addition salt of Ac-PHSCN-NH 2 is formed from methanesulfonic acid, acetic acid, glycolic acid, (+) camphorsulfonic acid, mandeleic acid, salicyclic acid, succinic acid or combinations thereof.
  • Exemplary inorganic acids include hydrofluoric acid, perchloric acid, hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid, phosphorous acid, hydroiodic acid, chloric acid, thiocyanic acid, hypophosphorous acid, nitrous acid, cyanic acid, chromic acid, sulfurous acid, and hydrazoic acid.
  • Other inorganic acids are known to those of skill in the art.
  • the acid addition salt OfAc-PHSCN-NH 2 is formed from hydrobromic acid, nitric acid, hydrochloric acid, phosphoric acid or combinations thereof.
  • the acid addition salt of Ac-PHSCN-NH 2 is formed from hydrochloric acid.
  • the acid addition salts OfAc-PHSCN-NH 2 may be made any conventional method known to those of skill in the art. These methods include saturating solutions OfAc-PHSCN-NH 2 with gaseous acids, adding solutions of acids to solutions OfAc-PHSCN-NH 2 , etc.
  • an acid addition salt OfAc-PHSCN-NH 2 is made by adding slightly more than 1 equivalent ⁇ e.g., 1.05 equivalent) of the acid to a solution OfAc-PHSCN-NH 2 dissolved in distilled water. The acid addition salt is typically isolated as a solid from the aqueous mixture.
  • the acid addition salt OfAc-PHSCN-NH 2 is generally, considerably more stable than the free base in both the solid and solution phase.
  • the acid addition salt is believed to prevent oxidative dimerization of Ac-PHSCN-NH 2 mediated by cysteine.
  • Acid addition salts which prevent degradation of Ac-PHSCN-NH 2 are formed from, for example, methanesulfonic acid, acetic acid, glycolic acid, sulfuric acid, (+) camphorsulfonic acid, mandeleic acid, salicyclic acid, succinic acid, hydrobromic acid, hydrochloric acid, nitric acid and phosphoric acid.
  • the formulation of the present invention preferably comprises one or more reducing agent, such as dithiothreitol (DTT) at a low concentration, preferably not exceeding about 10 mM, ⁇ -mercaptoethanol, glutathione (GSH) or other Cys-containing reducing agents.
  • DTT dithiothreitol
  • GSH glutathione
  • Other non-thiol containing anti-oxidants such as metal binding compounds (EDTA, EGTA) that are biocompatible may also be used.
  • Another embodiment of the invention is directed to formulations that increase half-life of the peptide after parenteral injection.
  • examples of these include formulation of Ac-PHSCN-NH 2 with a cyclodextrins, cremaphor or liposomal formulations.
  • Cyclodextrin refers to a cyclic molecule containing six or more cc-D-glucopyranose units linked at the 1,4 positions by ⁇ linkages as in amylose. ⁇ -Cyclodextrin or cycloheptaamylose contains seven oc-D-glucopyranose units.
  • cyclodextrin also includes cyclodextrin derivatives such as hydroxypropyl and sulfobutyl ether cyclodextrins. Such derivatives are described for example, in U.S. Patents No. 4,727,064 and 5,376,645.
  • cyclodextrin is hydroxypropyl ⁇ -cyclodextrin (H ⁇ CD) having a degree of substitution of from about 4.1-5.1 as measured by Fourier Transform Infrared Spectroscopy.
  • H ⁇ CD hydroxypropyl ⁇ -cyclodextrin
  • the Ac-PHSCN-NH 2 or derivative is formulated in an aqueous solution containing a cyclodextrin.
  • the peptide of this invention is formulated as a lyophilized powder containing a cyclodextrin or as a sterile powder containing a cyclodextrin.
  • the cyclodextrin is (H ⁇ CD) or sulfobutyl ether ⁇ -cyclodextrin; more preferably, the cyclodextrin is hydroxypropyl- ⁇ -cyclodextrin.
  • the cyclodextrin will comprise about 1 - 25% (w/w), preferably, about 2 - 10%, more preferably, about 4 - 6%, of the formulation.
  • the weight ratio of the cyclodextrin to the peptide will preferably be from about 1:1 to about 10:1.
  • Ac-PHSCN-NH 2 or its derivative is formulated to render the peptide orally or intranasally bioavailable.
  • polyols such as mannitol, which may act as stabilizers, and further help the bulk powder form a cake.
  • the polyol has at least three hydroxyl groups, and may be in the form of a mixture of two or more polyols.
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to, or following, lyophilization and reconstitution. Alternatively, sterility, of the entire mixture may be accomplished by autoclaving the ingredients prior to addition of the peptide at about 12O 0 C for about 30 minutes, for example. After the peptide, lyoprotectant and other optional components are mixed together, the formulation is lyophilized. Many different freeze-drivers are available for this purpose such as Hull50TM (Hull, USA) or GT20TM (Leybold-Heraeus, Germany) freeze-dryers.
  • Freeze-drying is accomplished by freezing the formulation and subsequently subliming ice from the frozen content at a temperature suitable for primary drying. Under this condition, the product temperature is below the eutectic point or the collapse temperature of the formulation. Typically, the shelf temperature for the primary drying will range from about -30 to 25°C (provided the product remains frozen during primary dying) at a suitable pressure, ranging typically from about 50 to 250 mTorr.
  • the formulation, size and type of the container holding the sample (e.g., glass vial) and the volume of liquid will mainly dictate the time required for drying, which can range from a few hours to several days (e.g., 40-60 hrs).
  • a secondary drying stage may be carried out at about 0-40°C, depending primarily on the type and size of container and nature of the peptide.
  • the shelf temperature throughout the entire water removal phase of lyophilization may be from about 15- 30°C. (e.g., about 2O 0 C).
  • the time and pressure required for secondary drying will be that which produces a suitable lyophilized cake, dependent e.g., on the temperature and other parameters.
  • the secondary drying time is dictated by the desired residual moisture level in the product and typically takes at least about 5 hours (e.g., 10-15 hours).
  • the pressure may be the same as that employed during the primary drying step. Freeze-drying conditions can be varied depending on the formulation and vial size.
  • the container in this instance may, for example, be a 3, 5, 10, 20, 50 or 100 cc vial.
  • lyophilization will result in a lyophilized formulation in which the moisture content thereof is less than about 5%, preferably less than about 3%.
  • the lyophilized formulation may be reconstituted with a diluent such that the peptide concentration in the reconstituted formulation is at least 10 mg/mL, for example from about 10 mg/mL to about 1000 mg/mL, more preferably from about 50 mg/mL to about 500 mg/mL, and most preferably from about 100 mg/mL to about 500 mg/mL.
  • a diluent such that the peptide concentration in the reconstituted formulation is at least 10 mg/mL, for example from about 10 mg/mL to about 1000 mg/mL, more preferably from about 50 mg/mL to about 500 mg/mL, and most preferably from about 100 mg/mL to about 500 mg/mL.
  • the peptide concentration in the reconstituted formulation is significantly higher than that in the pre-lyophilized formulation.
  • the peptide concentration in the reconstituted formulation may be about 2-40 times, preferably 3-10 times and most preferably 3-6 times (e.g., at least three fold or at least four fold) that of the pre-lyophilized formulation.
  • Reconstitution generally takes place at a temperature of about 25°C to ensure complete hydration, although other temperatures may be employed as desired.
  • the time required for reconstitution will depend, for example, on the type of diluent, amount of excipient(s) and peptide.
  • exemplary diluents include sterile water, bacteriostatic water for injection (BWFI), a pH buffered solution (e.g. phosphate-buffered saline, PBS), sterile saline solution, Ringer's solution or dextrose solution.
  • BWFI bacteriostatic water for injection
  • PBS phosphate-buffered saline
  • sterile saline solution e.g. phosphate-buffered saline
  • Ringer's solution or dextrose solution e.g. phosphate-buffered saline
  • the diluent optionally contains a preservative. Exemplary preservatives have been described above,
  • the amount of preservative employed is determined by assessing different preservative concentrations for compatibility with the peptide and preservative efficacy testing.
  • the preservative is an aromatic alcohol (such as benzyl alcohol)
  • it can be present in an amount from about 0.1-2.0% and preferably from about 0.5-1.5%, but most preferably about 1.0-1.2%.
  • the reconstituted formulation has less than 6000 particles per vial which are
  • an article of manufacture which contains the lyophilized formulation of the present invention and provides instructions for its reconstitution and/or use.
  • the article of manufacture comprises a container. Suitable containers include, for example, bottles, vials (e.g. dual chamber vials), syringes (such as dual chamber syringes) and test tubes.
  • the container may be formed from a variety of materials such as glass or plastic.
  • the container holds the lyophilized formulation and a label on, or associated with, the container may indicate directions for reconstitution and/or use.
  • the label may indicate that the lyophilized formulation is reconstituted to peptide concentrations as described above.
  • the label may further indicate that the formulation is useful or intended for subcutaneous administration.
  • the container holding the formulation may be a multi-use vial, which allows for repeat administrations (e.g., from 2-6 administrations) of the reconstituted formulation.
  • the article of manufacture may further comprise a second container comprising a suitable diluent (e.g., BWFI). Upon mixing of the diluent and the lyophilized formulation, the final protein concentration in the reconstituted formulation will generally be at least 50 mg/mL.
  • the article of manufacture may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • Therapeutic kits may have a single container which contains the formulation of the Ac-PHSCN-NH 2 pharmaceutical compositions with or without other components (e.g., other compounds or pharmaceutical compositions of these other compounds) or may have distinct container for each component.
  • therapeutic kits of the invention include a formulation of Ac-PHSCN-NH 2 or an acid addition salt thereof as disclosed herein packaged for use in combination with the co-administration of a second compound (such as a chemotherapeutic agent, a natural product, a hormone or antagonist, a anti-angiogenesis agent or inhibitor, a apoptosis-inducing agent or a chelator) or a pharmaceutical composition thereof.
  • a second compound such as a chemotherapeutic agent, a natural product, a hormone or antagonist, a anti-angiogenesis agent or inhibitor, a apoptosis-inducing agent or a chelator
  • the components of the kit may be pre-complexed or each component may be in a separate distinct container prior to administration to a patient.
  • the components of the kit may be provided in one or more liquid solutions, preferably, an aqueous solution, more preferably, a sterile aqueous solution.
  • the components of the kit may also be provided as solids, which may be converted into liquids by addition of suitable solvents, which are preferably provided in another distinct container.
  • the container of a therapeutic kit may be a vial, test tube, flask, bottle, syringe, or any other means of enclosing a solid or liquid.
  • the kit will contain a second vial or other container, which allows for separate dosing.
  • the kit may also contain another container for a pharmaceutically acceptable liquid.
  • a therapeutic kit will contain apparatus (e.g., one or more needles, syringes, eye droppers, pipette, etc.), which enables administration of the agents of the invention which are components of the present kit.
  • the present formulation is one that is suitable for administration of the peptide by any acceptable route such a oral (enteral), subcutaneous, intramuscular, intravenous transdermal, Administration may be by infusion pump. All modes and routes of administration disclosed by Ternansky et al., supra, are understood to be useful with the present formulations and are incorporated by reference as if written here in full.
  • the present invention is also directed to formulations of the peptide suitable for administration by inhalation. Many drugs currently administered by inhalation come primarily as liquid or solid aerosol particles of respirable size.
  • biodegradable carriers such as poly(lactide-co-glycolides
  • biotherapeutic proteins and peptides have been developed for biotherapeutic proteins and peptides (Liu, R. et al., Biotechnol. Bioeng., 57:177-184 (1991).
  • Most therapeutic peptides are poorly absorbed through biologic membranes even upon formulation with penetration enhancer.
  • drug dosimetry is increased by orders of magnitude to achieve minimum systemic concentrations required for efficacy.
  • the drug product is formulated with exotic absorption promoters in order to improve permeability across an absorption barrier, often leading to toxic consequences.
  • the mode of drug administration to the body has also gradually expanded from oral and parenteral to transdermal, rectal and the pulmonary routes of administration, i.e., nose and lung.
  • Success with these drug delivery approaches have been mixed due to lack of acceptance of the newer, complex molecules that must be used for treating difficult diseases, e.g., infections, malignancies, cardiovascular, endocrine, neurologic diseases, and a variety of diseases of immunological compromise.
  • the present invention exploits the existence of a fluid propelled formulation system comprising the peptide (drug) that is stable and protected by a rate-limiting carrier, easily manufactured, and therapeutically effective when administered as fluid dispersed particles to the lung of a patient.
  • the present invention includes formulations of the peptide for oral or nasal inhalation.
  • formulations include, but are not limited to, modulated release aerosol particles, and to medicinal, respirable aerosol particles comprising polysaccharide vesicles which are associated with, and, may form a part of, a construct with or entrap a selected medicament, here, the peptide, and provide slow release thereof, as disclosed in U.S. Pat. 6,551,578, incorporated by reference in its entirety.
  • the peptide is formulated so that it is suitable for administration by oral and nasal inhalation.
  • a stable, colloidal dispersion of the peptide in a fluid e.g., air, hydrocarbon gases, chlorofluorocarbon (CFC) propellants or non-CFC propellants, such as tetrafluoroethane (HFA- 134a) and heptafluoropropane (HFA-227) are intended.
  • a fluid e.g., air, hydrocarbon gases, chlorofluorocarbon (CFC) propellants or non-CFC propellants, such as tetrafluoroethane (HFA- 134a) and heptafluoropropane (HFA-227) are intended.
  • a fluid e.g., air, hydrocarbon gases, chlorofluorocarbon (CFC) propellants or non-CFC propellants, such as tetrafluoroethane (HFA- 134a) and heptafluoropropane (HFA-227) are intended.
  • CFC chlorofluor
  • the peptide is associated with the naturally occurring polysaccharide polymer to which it is destined to be combined.
  • association in this context is meant that the peptide is present as a matrix or a part of a polymeric construct along with the polysaccharide polymer or is encapsulated as a microsphere in a polysaccharide polymer or in polysaccharide polymeric construct particle, or is on a surface of such particle, whereby a therapeutically effective amount or fraction (e.g., 95% or more) of the peptide is particulate.
  • the construct particles have a diameter of less than about 10 ⁇ m, and preferably less than about 5 ⁇ m, in order that the particles can be inhaled into the respiratory tract and/or lungs of the patient being treated.
  • a suitable polymeric construct is one which will incorporate therein or encapsulate the selected peptide and provide a controlled or modulated release of the medicament therefrom to the sites of action or application of the patient's body, e.g., from the lung to the local surrounding environment of the subject.
  • a suitable polysaccharide is a polymer selected from the group of an alginate salt, where the cation is, e.g., Li + , Na + , K + , Ca ++ , NH 3+ , NH 4+ etc., such as sodium alginate, calcium alginate, sodium-calcium alginate, ammonium alginate, sodium-ammonium alginate, or calcium-ammonium alginate.
  • a preferred alginate modulating releasing agent is ammonium calcium alginate. These materials are typically used in injectable implants and microsphere preparations for controlled release.
  • a commercial form of ammonium calcium alginate is KeltoseTM, manufactured and distributed by International Specialty Products (Wayne, NJ).
  • alginate means alginic acid, or any of its salts; or other naturally occurring polysaccharide or carbohydrate based polymers such as gum arabic, pectin, galacturonic acid, gum karaya; gum Benjamin, plantago ovata gum; agar; carrageenan; cellulose; gelatin; or a mixture of any of the foregoing polymers.
  • Alginates are pharmaceutical excipients generally regarded as safe and used to prepare a variety of well documented pharmaceutical systems (U.S. Pats. No. 6,166,084; 6,166,043; 6,166,042; 6,166,004; and 6,165,615). Alginates are naturally occurring polymers comprising polysaccharide chains.
  • polysaccharide polymer e.g., an alginate salt
  • the polysaccharide polymer is typically present in the resultant controlled-release formulation in an amount ranging from about 0.000001% to about 10% by weight of the total weight of the formulation.
  • the therapeutic peptide is present in the inventive polymer construct in a therapeutically effective amount, that is, an amount such that the peptide can be incorporated into an aerosol formulation such as a dispersion aerosol, via oral or nasal inhalation, and cause its desired therapeutic effect, preferably with one dose, or through several doses.
  • a therapeutically effective amount that is, an amount such that the peptide can be incorporated into an aerosol formulation such as a dispersion aerosol, via oral or nasal inhalation, and cause its desired therapeutic effect, preferably with one dose, or through several doses.
  • Liposomes may be phospholipid or non-phospholipid based.
  • transwells are coated with type I collagen (50 ⁇ g/mL) by adding 200 ⁇ L of the collagen solution per transwell, then incubating overnight at 37°C.
  • the transwells are assembled in a 24- well plate and a chemoattractant (e.g., FGF-2) is added to the bottom chamber in a total volume of 0.8 mL media.
  • ECs such as human umbilical vein endothelial cells (HUVEC), which have been detached from monolayer culture using trypsin, are diluted to a final concentration of about 10 6 cells/mL with serum-free media and 0.2 mL of this cell suspension is added to the upper chamber of each transwell.
  • a chemoattractant e.g., FGF-2
  • Salts of Ac-PHSCN-NH 2 may be added to both the upper and lower chambers and the migration is allowed to proceed for 5 hrs in a humidified atmosphere at 37°C.
  • the transwells are removed from the plate stained using DiffQuik ® .
  • Cells which did not migrate are removed from the upper chamber by scraping with a cotton swab and the membranes are detached, mounted on slides, and counted under a high-power field (40Ox) to determine the number of cells migrated.
  • Matrigel® is a reconstituted basement membrane containing type IV collagen, laminin, heparan sulfate proteoglycans such as perlecan, which bind to and localize bFGF, vitronectin as well as transforming growth factor- ⁇ (TGF ⁇ , urokinase-type plasminogen activator (uPA), tissue plasminogen activator (tPA) and the serpin known as plasminogen activator inhibitor type 1 (PAI-I) (Chambers et al, Cane. Res. 1995, 55:1578-1585,). It is accepted in the art that results obtained in this assay for compounds which target extracellular receptors or enzymes are predictive of the efficacy of these compounds in vivo (Rabbani et al, Int. J. Cancer 1995, 63: 840-845).
  • Such assays employ transwell tissue culture inserts.
  • Invasive cells are defined as cells which are able to traverse through the Matrigel® and upper aspect of a polycarbonate membrane and adhere to the bottom of the membrane.
  • Transwells (Costar) containing polycarbonate membranes (8.0 ⁇ m pore size) are coated with Matrigel® (Collaborative Research), which has been diluted in sterile PBS to a final concentration of 75 ⁇ g/mL (60 ⁇ L of diluted Matrigel® per insert), and placed in the wells of a 24-well plate.
  • the membranes are dried overnight in a biological safety cabinet, then rehydrated by adding 100 ⁇ L of DMEM containing antibiotics for 1 hour on a shaker table.
  • the DMEM is removed from each insert by aspiration and 0.8 mL of DMEM/10 % FBS/antibiotics is added to each well of the 24-well plate such that it surrounds the outside of the transwell ("lower chamber”).
  • Fresh DMEM/ antibiotics (lOO ⁇ L), human Glu-plasminogen (5 ⁇ g/mL), and any inhibitors to be tested are added to the top, inside of the transwell ("upper chamber”).
  • the cells which are to be tested are trypsinized and resuspended in DMEM/antibiotics, then added to the top chamber of the transwell at a final concentration of 800,000 cells/mL.
  • the final volume of the upper chamber is adjusted to 200 ⁇ L.
  • the assembled plate is then incubated in a humid 5% CO 2 atmosphere for 72 hours. After incubation, the cells are fixed and stained using DiffQuik® (Giemsa stain) and the upper chamber is then scraped using a cotton swab to remove the Matrigel® and any cells which did not invade through the membrane.
  • the membranes are detached from the transwell, e.g., using an X-acto ® blade, mounted on slides using Permount ® and cover-slips, then counted under a high-powered (40Ox) field. An average of the cells invaded is determined from 5-10 fields counted and plotted as a function of peptide concentration.
  • Endothelial cells for example, human umbilical vein endothelial cells (HUVEC) or human microvascular endothelial cells (HMVEC) which can be prepared or obtained commercially, are mixed at a concentration of 2 x 10 5 cells/mL with fibrinogen (5mg/mL in phosphate buffered saline (PBS) in a 1:1 (v/v) ratio. Thrombin is added (5 units/ mL final concentration) and the mixture is immediately transferred to a 24-well plate (0.5 mL per well).
  • HMVEC human umbilical vein endothelial cells
  • HMVEC human microvascular endothelial cells
  • VEGF and bFGF are added to the wells (each at 5 ng/mL final concentration) along with the test compound.
  • the cells are incubated at 37°C in 5% CO 2 for 4 days at which time the cells in each well are counted and classified as either rounded, elongated with no branches, elongated with one branch, or elongated with 2 or more branches. Results are expressed as the average of 5 different wells for each concentration of compound.
  • angiogenic inhibitors cells remain either rounded or form undifferentiated tubes ⁇ e.g. 0 or 1 branch). This assay is recognized in the art to be predictive of angiogenic (or anti-angiogenic) efficacy in vivo (Min et at, Cancer Res. 1996, 56: 2428-2433,).
  • endothelial cell tube formation is observed when endothelial cells are cultured on Matrigel® (Schnaper et at, J. Celt Physiol. 1995, 165: 107-118). Endothelial cells (10 4 cells/well) are transferred onto Matrigel®-coated 24-well plates and tube formation is quantitated after 48 hrs. Inhibitors are tested by adding them either at the same time as the endothelial cells or at various time points thereafter. Tube formation can also be stimulated by adding (a) angiogenic growth factors such as bFGF or VEGF, (b) differentiation stimulating agents ⁇ e.g.,. PMA) or (c) a combination of these.
  • this assay models angiogenesis by presenting to the endothelial cells a particular type of basement membrane, namely the layer of matrix which migrating and differentiating endothelial cells might be expected to first encounter.
  • the matrix components found in Matrigel® (and in basement membranes in situ) or proteolytic products thereof may also be stimulatory for endothelial cell tube formation which makes this model complementary to the fibrin gel angiogenesis model previously described (Blood et al, Biochim. Biophys. Acta 1990, i 032:89-118; Odedrat al, Pharmac. Ther. 1991, 49:111-124,).
  • the ability of the compounds of the invention to inhibit the proliferation of EC's may be determined in a 96-well format.
  • Type I collagen (gelatin) is used to coat the wells of the plate (0.1-1 mg/mL in PBS, 0.1 mL per well for 30 minutes at room temperature). After washing the plate (3x w/PBS), 3-6,000 cells are plated per well and allowed to attach for 4 hrs (37°C/5% CO 2 ) in Endothelial Growth Medium (EGM; Clonetics ) or M199 media containing 0.1-2% FBS.
  • the media and any unattached cells are removed at the end of 4 hrs and fresh media containing bFGF (1-10 ng/mL) or VEGF (1-10 ng/rnL) is added to each well.
  • Compounds to be tested are added last and the plate is allowed to incubate (37°C/5% CO 2 ) for 24-48 hrs.
  • MTS Promega
  • the absorbance at 490nm, which is proportional to the cell number, is then measured to determine the differences in proliferation between control wells and those containing test compounds.
  • a similar assay system can be set up with cultured adherent tumor cells. However, collagen may be omitted in this format.
  • Tumor cells e.g., 3,000-10,000/well
  • Serum free medium is then added to the wells, and the cells are synchronized for 24 hrs.
  • Medium containing 10% FBS is then added to each well to stimulate proliferation.
  • Compounds to be tested are included in some of the wells. After 24 hrs, MTS is added to the plate and the assay developed and read as described above.
  • the ability of the compounds of the invention to promote apoptosis of EC's may be determined by measuring activation of caspase-3.
  • Type I collagen gelatin
  • 5xlO 5 ECs are seeded in EGM containing 10% FBS.
  • the cells are harvested after 6 hours, cell lysates prepared in 1% Triton and assayed using the EnzChek®Caspase-3 Assay Kit #1 (Molecular Probes) according to the manufactures' instructions.
  • Neovascularization is assessed at 5 and 7 days after implantation. On day 7, animals are anesthetized and infused with a dye such as colloidal carbon to stain the vessels. The animals are then euthanized, the corneas fixed with formalin, and the corneas flattened and photographed to assess the degree of neovascularization. Neovessels may be quantitated by imaging the total vessel area or length or simply by counting vessels. Matrigel® Plug Assay
  • This assay is performed essentially as described by Passaniti et at, 1992, Lab Invest. 67:519- 528. Ice-cold Matrigel® (e.g., 500 ⁇ L) (Collaborative Biomedical Products, Inc., Bedford, MA) is mixed with heparin (e.g., 50 ⁇ g/ml), FGF-2 (e.g., 400 ng/ml) and the compound to be tested. In some assays, bFGF may be substituted with tumor cells as the angiogenic stimulus.
  • the Matrigel® mixture is injected subcutaneously into 4-8 week-old athymic nude mice at sites near the abdominal midline, preferably 3 injections per mouse.
  • the injected Matrigel® forms a palpable solid gel. Injection sites are chosen such that each animal receives a positive control plug (such as FGF-2 + heparin), a negative control plug (e.g., buffer + heparin) and a plug that includes the compound being tested for its effect on angiogenesis, e.g., (FGF-2 + heparin + compound).
  • AU treatments are preferably run in triplicate. Animals are sacrificed by cervical dislocation at about 7 days post injection or another time that may be optimal for observing angiogenesis. The mouse skin is detached along the abdominal midline, and the Matrigel® plugs are recovered and scanned immediately at high resolution. Plugs are then dispersed in water and incubated at 37°C overnight.
  • Hemoglobin (Hb) levels are determined using Drabkin's solution (e.g., obtained from Sigma) according to the manufacturers' instructions.
  • the amount of Hb in the plug is an indirect measure of angiogenesis as it reflects the amount of blood in the sample.
  • animals may be injected prior to sacrifice with a 0.1 ml buffer (preferably PBS) containing a high molecular weight dextran to which is conjugated a fluorophore.
  • PBS 0.1 ml buffer
  • the amount of fluorescence in the dispersed plug determined fluorimetrically, also serves as a measure of angiogenesis in the plug.
  • CD31 is "platelet-endothelial cell adhesion molecule or PECAM"
  • PECAM platelet-endothelial cell adhesion molecule
  • This assay is performed essentially as described by Nguyen et at, Microvascular Res. 1994, 47:31-40.
  • a mesh containing either angiogenic factors (bFGF) or tumor cells plus inhibitors is placed onto the CAM of an 8-day old chick embryo and the CAM observed for 3-9 days after implantation of the sample.
  • Angiogenesis is quantitated by determining the percentage of squares in the mesh which contain blood vessels.
  • tumor cells for example 1-5 x 10 6 cells of the 3LL Lewis lung carcinoma or the rat prostate cell line MatLyLu, are mixed with Matrigel® and then injected into the flank of a mouse following the protocol described in Sec. B., above.
  • a mass of tumor cells and a powerful angiogenic response can be observed in the plugs after about 5 to 7 days.
  • the anti-tumor and anti- angiogenic action of a compound in an actual tumor environment can be evaluated by including it in the plug.
  • Measurement is then made of tumor weight, Hb levels or fluorescence levels (of a dextran-fluorophore conjugate injected prior to sacrifice).
  • the plugs are first homogenize with a tissue homogenizer.
  • Nude mice are inoculated with MDA-MB-231 cells (human breast carcinoma) and Matrigel® (10 6 cells in 0.2mL) s.c. in the right flank of the animals.
  • the tumors are staged to 200 mm 3 and then treatment with a test composition is initiated (lOO ⁇ g/animal/day given q.d. IP).
  • Tumor volumes are obtained every other day and the animals are sacrificed after 2 weeks of treatment.
  • the tumors are excised, weighed and paraffin embedded. Histological sections of the tumors are analyzed by H and E, anti-CD31, Ki-67, TUNEL, and CD68 staining.
  • Xenograft Model of Metastasis The compounds of the invention are also tested for inhibition of late metastasis using an experimental metastasis model (Crowley et al, Proc. Natl. Acad. ScL USA 1993, 905021-5025). Late metastasis involves the steps of attachment and extravasation of tumor cells, local invasion, seeding, proliferation and angiogenesis.
  • Human prostatic carcinoma cells PC-3) transfected with a reporter gene, preferably the green fluorescent protein (GFP) gene, but as an alternative with a gene encoding the enzymes chloramphenicol acetyl-transferase (CAT), luciferase or LacZ, are inoculated into nude mice.
  • a reporter gene preferably the green fluorescent protein (GFP) gene
  • This approach permits utilization of either of these markers (fluorescence detection of GFP or histochemical colorimetric detection of enzymatic activity) to follow the fate of these cells.
  • Cells are injected, preferably iv, and metastases identified after about 14 days, particularly in the lungs but also in regional lymph nodes, femurs and brain. This mimics the organ tropism of naturally occurring metastases in human prostate cancer.
  • GFP-expressing PC-3 cells (10 6 cells per mouse) are injected iv into the tail veins of nude (nu/nu) mice. Animals are treated with a test composition at lOO ⁇ g/animal/day given q.d. IP.
  • Single metastatic cells and foci are visualized and quantitated by fluorescence microscopy or light microscopic histochemistry or by grinding the tissue and quantitative colorimetric assay of the detectable label.
  • the rat syngeneic breast cancer system employs Mat BUI rat breast cancer cells (Xing et al,
  • Tumor cells for example, about 10 6 suspended in 0.1 mL PBS, are inoculated into the mammary fat pads of female Fisher rats.
  • a 14-day Alza osmotic mini-pump is implanted intraperitoneally to dispense the test compound.
  • the compound is dissolved in PBS (e.g., 200 mM stock), sterile filtered and placed in the minipump to achieve a release rate of about 4 mg/kg/day.
  • Control animals receive vehicle (PBS) alone or a vehicle control peptide in the minipump. Animals are sacrificed at about day 14.
  • This tumor line arose spontaneously as carcinoma of the lung in a C57BL/6 mouse (Malave et al, J. Nafl. Cane. Inst. 1979, 62:83-88). It is propagated by passage in C57BL/6 mice by subcutaneous (sc) inoculation and is tested in semiallogeneic C57BL/6 x DBA/2 F 1 mice or in allogeneic C3H mice. Typically six animals per group for subcutaneously (sc) implant, or ten for intramuscular (im) implant are used. Tumor may be implanted sc as a 2-4 mm fragment, or im or sc as an inoculum of suspended cells of about 0.5-2 x 10 6 -cells. Treatment begins 24 hours after implant or is delayed until a tumor of specified size (usually approximately 400 mg) can be palpated. The test compound is administered ip daily for 11 days
  • Test mice are male C57BL/6 mice, 2-3 months old. Following sc, im, or intra-footpad implantation, this tumor produces metastases, preferentially in the lungs. With some lines of die tumor, the primary tumor exerts anti-metastatic effects and must first be excised before study of the metastatic phase (see also U.S. Patent No. 5,639,725).
  • Viability of 3LL cells prepared in this way is generally about 95-99%.
  • Viable tumor cells (3 x 10 4 - 5 x 10 6 ) suspended in 50 ⁇ l PBS are injected sc, either in the dorsal region or into one hind foot pad of C57BL/6 mice. Visible tumors appear 3-4 days after dorsal sc injection of 10 6 cells. The day of tumor appearance and the diameters of established tumors are measured every two days. The treatment is given as one to five doses of peptide or analogue per week. In another embodiment, the peptide is delivered by osmotic minipump.
  • mice are randomized into two groups: (1) primary tumor is completely excised; or (2) sham surgery is performed and the tumor is left intact. Although tumors from 500-3000 mm 3 inhibit growth of metastases, 1500 mm 3 is the largest size primary tumor that can be safely resected with high survival and without local regrowth. The phenomenon of acceleration of metastatic growth following excision of local tumors had been repeatedly observed (see, for example, U. S. Pat. No. 5,639,725). These observations have implications for the prognosis of patients who undergo cancer surgery. After 21 days, all mice are sacrificed and autopsied.
  • Lungs are removed, weighed and fixed in Bouin's solution and the number of visible metastases is recorded as is the diameters of the metastases. On the basis of the recorded diameters, one calculates the volume of each metastasis. To determine the total volume of metastases per lung, the mean number of visible metastases is multiplied by the mean volume.
  • mice with footpad tumors when tumors reach about 8-10 mm in diameter, mice are randomized into two groups: (1) legs with tumors are amputated after ligation above the knee joints; or (2) mice are left intact as nonamputated tumor-bearing controls. (Amputation of a tumor-free leg in a tumor-bearing mouse has no known effect on subsequent metastasis, ruling out possible effects of anesthesia, stress or surgery.) Mice are killed 10-14 days after amputation. Metastases are evaluated as above.
  • An Ac-PHSCN-NH 2 salt, or pharmaceutical compositions thereof, will generally be used in an amount effective to achieve the intended purpose.
  • the Ac-PHSCN-NH 2 salts which may be in pharmaceutical compositions, are administered or applied in a therapeutically effective amount.
  • the amount of a Ac-PHSCN-NH 2 salt that will be effective in the treatment of a particular disorder or condition disclosed herein will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques known in the art.
  • in vitro or in vivo assays may optionally be employed to help identify optimal doses or dose ranges.
  • the amount of an Ac-PHSCN-NH 2 salt administered will, of course, be dependent on, among other factors, the subject being treated, the weight of the subject, the severity of the affliction, the manner of administration and the judgment of the prescribing physician.
  • the dosage may be delivered in a pharmaceutical composition by a single or multiple administrations, or by controlled release. Dosing may be repeated intermittently, may be performed alone or in combination with other drugs and may be continued as long as required for effective treatment of the disease or disorder.
  • Suitable dosage ranges for oral administration are dependent on the potency of the drug, but are generally 0.001 mg to 200 mg, preferably 0.01 mg to 50 mg, more preferably, 0.1 to 50 mg, of a compound of the invention per kilogram body weight.
  • Suitable dose ranges for i.v. administration are about 0.01 mg to about 100 mg per kg body weight.
  • Suitable dosage ranges for intranasal administration are generally 0.01 mg/kg body weight to 50 mg/kg body weight or 0.10 mg/kg body weight to 10 mg/kg body weight.
  • Suppositories generally contain about 0.01 mg to about 50 mg of the compound of the invention per kg body weight and comprise about 0.5% to about 10% by weight the active ingredient.
  • Recommended dosages for intradermal, intramuscular, intraperitoneal, subcutaneous, epidural, sublingual or intracerebral administration are in the range of about 0.001 mg to about 200 mg per kg body weight.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. Such animal models and systems are well-known in the art.
  • the dose administered is not based on body weight, but is an absolute amount, for example, in the range of 1 mg to 1 g per dose.
  • the dose is 10 -750 mg per dose, e.g., 20 mg, 100 mg, or 600 mg per dose.
  • the dose is administered from one to several (e.g., 2, 3, 4, or 7) times per week.
  • the Ac-PHSCN-NH 2 salts are preferably assayed in vitro and in vivo, as described above for the desired therapeutic or prophylactic activity, prior to use in humans.
  • in vitro assays can be used to determine whether administration of an Ac-PHSCN-NH 2 salt or a combination of Ac- PHSCN-NH 2 salts is preferred for treating diseases characterized by aberrant angiogenesis or vascularization.
  • Safety and efficacy of the Ac-PHSCN-NH 2 salts may be demonstrated using animal model systems.
  • a therapeutically effective dose of a Ac-PHSCN-NH 2 salt described herein will provide therapeutic benefit without causing substantial toxicity.
  • Toxicity of Ac-PHSCN- NH 2 salts may be determined using standard pharmaceutical procedures and may be readily ascertained by the skilled artisan.
  • the "therapeutic index" ratio between a toxic and a therapeutic dose
  • the preferred dose of an Ac-PHSCN-NH 2 salt described herein will preferably result in a range of circulating concentrations of the agent that are effective but with little or no toxicity.
  • Rink Amide AM resin (Novabiochem) was treated with 20% piperidine in DMF (1 mL per 100 mg of resin) for three minutes with nitrogen agitation and the reaction mixture was filtered and washed once with DMF. This step was repeated an additional two times. The resin was washed three times with DMF and three times with dichloromethane. Fmoc-Asn(trt)-OH (3 eq), HBTU (3 eq), and HOBt (3 eq) were dissolved in DMF (1 mL per 100 mg of resin) and added to the above resin, followed by the addition of N-methylmorpholine ( ⁇ MM) (6 eq) and the mixture was agitated for 1 hour.
  • ⁇ MM N-methylmorpholine
  • the crude peptide dissolved in a minimum amount of methanol and water, was purified by preparative reverse phase HPLC (Beckman) with a Phenomenex Synergi hydro-RP C18 column (250mm x 21.2 mm).
  • the peptide was eluted using a gradient from 3-100% B over 30 min with a flow rate of 20 mL/min, where solvent A was water containing 0.1% trifluoroacetic acid and solvent B was acetonitrile containing 0.1% trifluoroacetic acid. Detection was at 220 nm.
  • Accelerated Stability was tested by storage of the solution for 4 day at 40°C and 34 days at 55°C. Accelerated stability experiments are performed using extreme conditions of temperature and/or humidity to force degradation and to quickly test the relative stability of a particular formulation (typical storage conditions are 22°, 4°, or -20°C for most drugs). This data then provides the ability to select a formulation that will most likely be the most stable from a number of test formulations. After reconstitution with water each formulation was examined by reverse phase HPLC using a standard mobile phases such as water/methanol or water/acetonitrile. This methodology allows the separation of Ac-PHSCN-NH 2 from Ac-PHSCN-NH 2 degradation products such as fragments of Ac-PHSCN-NH 2 or the disulfide-bonded dimer.
  • Purity was calculated as the area under the Ac-PHSCN-NH 2 peak as a percentage of the integrated area of entire chromatogram.
  • the relative amount OfAc-PHSCN-NH 2 dimer was calculated as the area under the dimer peak as a percentage of the integrated area of entire chromatogram.
  • composition of solution 5 is 50mg/mL of the peptide, 50mg/mL glycine, 5OmM citrate, pH 5.0 , lyophilized (ImL). 2. Robust performance in the lyophilizer, uniform appearance with no collapse in any sample.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Dermatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
EP06720021A 2005-02-01 2006-02-01 Zusammensetzungen mit antiangiogenem phscn-peptid Pending EP1843779A2 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US64839105P 2005-02-01 2005-02-01
PCT/US2006/003461 WO2006083906A2 (en) 2005-02-01 2006-02-01 Compositions containing the anti-angiogenic phscn-peptide

Publications (1)

Publication Number Publication Date
EP1843779A2 true EP1843779A2 (de) 2007-10-17

Family

ID=36581633

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06720021A Pending EP1843779A2 (de) 2005-02-01 2006-02-01 Zusammensetzungen mit antiangiogenem phscn-peptid

Country Status (6)

Country Link
EP (1) EP1843779A2 (de)
JP (1) JP2008528630A (de)
KR (1) KR20070100832A (de)
AU (1) AU2006210895A1 (de)
CA (1) CA2596255A1 (de)
WO (1) WO2006083906A2 (de)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0710529D0 (en) 2007-06-01 2007-07-11 Circassia Ltd Vaccine
SI2083856T1 (sl) 2007-08-15 2011-02-28 Circassia Ltd Peptidi za desenzibilizacijo proti alergijam
CN101503458B (zh) * 2008-02-04 2012-05-09 上海市第一人民医院 预防和治疗血管新生的小分子多肽及其应用
GB0821806D0 (en) * 2008-11-28 2009-01-07 Circassia Ltd Compositions with reduced dimer formation
WO2011066386A1 (en) * 2009-11-25 2011-06-03 Novetide, Ltd. Process for production of degarelix
JP6475714B2 (ja) * 2013-11-07 2019-02-27 ドクトア・アウグスト・ボルフ・ゲーエムベーハー・ウント・コンパニー・カー・ゲー−アルツナイミツテル 貯蔵安定性凍結乾燥トリペプチド製剤
KR20210045397A (ko) * 2018-07-11 2021-04-26 이뮤니티 파르마 엘티디. 펩타이드 화합물 및 이의 치료적 용도

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5140010A (en) * 1989-09-28 1992-08-18 Immunobiology Research Institute Stabilized aqueous formulations of thymopentin
WO1998022617A1 (en) * 1996-11-21 1998-05-28 The Regents Of The University Of Michigan Invasion-inducing agents and invasion-inhibitors for use in wound healing and cancer
PL377701A1 (pl) * 2002-11-25 2006-02-06 Attenuon, Llc Peptydy, które namierzają komórki nowotworowe i śródbłonkowe, kompozycje i ich zastosowania

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2006083906A2 *

Also Published As

Publication number Publication date
AU2006210895A1 (en) 2006-08-10
CA2596255A1 (en) 2006-08-10
KR20070100832A (ko) 2007-10-11
WO2006083906A3 (en) 2006-10-05
JP2008528630A (ja) 2008-07-31
WO2006083906A2 (en) 2006-08-10

Similar Documents

Publication Publication Date Title
EP1843779A2 (de) Zusammensetzungen mit antiangiogenem phscn-peptid
RU2518240C2 (ru) Композиция, на основе гидрофобных агентов и способ ее получения(варианты)
FI85335C (fi) Foerfarande foer framstaellning av lyofiliserad, farmaceutisk vaevnadsplasminogenaktivator(t-pa)-komposition.
US20090220435A1 (en) Tight junction modulating peptide components for enhancing mucosal delivery of therapeutic agents
CN107075574A (zh) 铁调素和微型铁调素类似物及其用途
KR100705997B1 (ko) 안정화된 hgf 동결건조제제 및 그 제조방법
EP3185880B1 (de) Verbesserte peptidyl-calcineurin-hemmer
US20100021527A1 (en) Collagen-related peptides and uses thereof and hemostatic foam substrates
JP6030201B2 (ja) α‐1プロテイナーゼインヒビターのための組成物、方法およびキット
AU2015233635B2 (en) Dry-powder peptide medicament
EP1042003A1 (de) Verabreichung über längere zeit wirksamer therapeutischen mittel durch bildung kovalenter bindungen in vivo
PT1196433E (pt) Derivados de peptidos ciclicos como inibidores da integrina clvbg
US20210228675A1 (en) Hypertrophic scar inhibiting composition
JP2011510043A (ja) 新規血液凝固因子インヒビター
CN111848736B (zh) 自组装多肽、制备方法、自组装多肽制剂及应用
AU771099B2 (en) Inhibitors of the integrin alphavbeta6
AU2006210627B2 (en) Acid addition salts of Ac-PHSCN-NH2
EP3333177B1 (de) Multitarget-verbindung mit antikoagulations- und thrombozytenaggregationshemmender wirkung, herstellungsverfahren dafür und verwendung davon
US6335320B1 (en) Method of treating fibrotic conditions
CN101784560A (zh) 凝血因子抑制剂
WO2023225619A2 (en) Delivery of focal adhesion kinase for treating vascular and/or capillary tissue injury
US20060148716A1 (en) Peptidic sulfonamides
MX2008000600A (es) Formulaciones para el suministro mucoso mejorado de pyy

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070801

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

17Q First examination report despatched

Effective date: 20071107

RIN1 Information on inventor provided before grant (corrected)

Inventor name: HARRIS, SCOTT

Inventor name: LI, MING

Inventor name: SCHRIER, JAY

Inventor name: HEIATI, HASHEM

Inventor name: MAZAR, ANDREW, P

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN